51
|
Ma Q, Gao S, Li C, Yao J, Xie Y, Jiang C, Yuan J, Fei K, Zhang P, Wang H, Li X. Cuproptosis and Serine Metabolism Blockade Triggered by Copper-Based Prussian Blue Nanomedicine for Enhanced Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406942. [PMID: 39676407 DOI: 10.1002/smll.202406942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/26/2024] [Indexed: 12/17/2024]
Abstract
Cuproptosis, a newly defined cell death process, represents a novel modality with significant therapeutic potential in cancer treatment. Nevertheless, the modest concentration and transient half-life of copper ions in the bloodstream constrain their efficient delivery into tumor cells. In this study, a copper-based prussian blue nanostructure loaded with serine metabolic inhibitor (NCT-503@Cu-HMPB) is constructed for selectively inducing cuproptosis combined with disrupting serine metabolism. Released within the tumor cells, NCT-503 is found to inhibit cellular serine metabolism and GSH production, ultimately causing metabolic dysfunction, redox imbalance, and increased the formation of Cu+ that disrupts mitochondrial respiration chain, inducing lipoylated protein dihydrolipoamide S-acetyltransferase (DLAT) aggregation and consequential iron-sulfur cluster protein loss, which leads to proteotoxic stress and ultimately results in cell death. The findings provide a novel paradigm for tumor therapy based on cuproptosis and metabolic reprogramming, offering prospects for the development of innovative nanotherapeutic platforms in the future.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Shanshan Gao
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Chaoyang Li
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Junjie Yao
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200010, P. R. China
| | - Yumeng Xie
- Shanghai JiaoTong University School of Medicine, Shanghai, 200025, P. R. China
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Jie Yuan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Ke Fei
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Xiaoguang Li
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| |
Collapse
|
52
|
Al-Faze R, Ahmed HA, El-Atawy MA, Zagloul H, Alshammari EM, Jaremko M, Emwas AH, Nabil GM, Hanna DH. Mitochondrial dysfunction route as a possible biomarker and therapy target for human cancer. Biomed J 2025; 48:100714. [PMID: 38452973 PMCID: PMC11743316 DOI: 10.1016/j.bj.2024.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
Mitochondria are vital organelles found within living cells and have signalling, biosynthetic, and bioenergetic functions. Mitochondria play a crucial role in metabolic reprogramming, which is a characteristic of cancer cells and allows them to ensure a steady supply of proteins, nucleotides, and lipids to enable rapid proliferation and development. Their dysregulated activities have been associated with the growth and metastasis of different kinds of human cancer, particularly ovarian carcinoma. In this review, we briefly demonstrated the modified mitochondrial function in cancer, including mutations in mitochondrial DNA (mtDNA), reactive oxygen species (ROS) production, dynamics, apoptosis of cells, autophagy, and calcium excess to maintain cancer genesis, progression, and metastasis. Furthermore, the mitochondrial dysfunction pathway for some genomic, proteomic, and metabolomics modifications in ovarian cancer has been studied. Additionally, ovarian cancer has been linked to targeted therapies and biomarkers found through various alteration processes underlying mitochondrial dysfunction, notably targeting (ROS), metabolites, rewind metabolic pathways, and chemo-resistant ovarian carcinoma cells.
Collapse
Affiliation(s)
- Rawan Al-Faze
- Department of Chemistry, Faculty of Science, Taibah University, Almadinah Almunawarah, Saudi Arabia
| | - Hoda A Ahmed
- Chemistry Department, Faculty of Science at Yanbu, Taibah University, Yanbu, Saudi Arabia; Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Mohamed A El-Atawy
- Chemistry Department, Faculty of Science at Yanbu, Taibah University, Yanbu, Saudi Arabia; Chemistry Department, Faculty of Science, Alexandria University, Ibrahemia, Alexandria, Egypt
| | - Hayat Zagloul
- Chemistry Department, Faculty of Science at Yanbu, Taibah University, Yanbu, Saudi Arabia
| | - Eida M Alshammari
- Department of Chemistry, College of Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Mariusz Jaremko
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs., King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Gehan M Nabil
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Demiana H Hanna
- Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt.
| |
Collapse
|
53
|
Wernhart S, Rassaf T. Exercise, cancer, and the cardiovascular system: clinical effects and mechanistic insights. Basic Res Cardiol 2025; 120:35-55. [PMID: 38353711 PMCID: PMC11790717 DOI: 10.1007/s00395-024-01034-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/21/2024] [Indexed: 03/05/2024]
Abstract
Cardiovascular diseases and cancer are the leading causes of death in the Western world and share common risk factors. Reduced cardiorespiratory fitness (CRF) is a major determinant of cardiovascular morbidity and cancer survival. In this review we discuss cancer- induced disturbances of parenchymal, cellular, and mitochondrial function, which limit CRF and may be antagonized and attenuated through exercise training. We show the impact of CRF on cancer survival and its attenuating effects on cardiotoxicity of cancer-related treatment. Tailored exercise programs are not yet available for each tumor entity as several trials were performed in heterogeneous populations without adequate cardiopulmonary exercise testing (CPET) prior to exercise prescription and with a wide variation of exercise modalities. There is emerging evidence that exercise may be a crucial pillar in cancer treatment and a tool to mitigate cardiotoxic treatment effects. We discuss modalities of aerobic exercise and resistance training and their potential to improve CRF in cancer patients and provide an example of a periodization model for exercise training in cancer.
Collapse
Affiliation(s)
- Simon Wernhart
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
54
|
Liang J, Vitale T, Zhang X, Jackson TD, Yu D, Jedrychowski M, Gygi SP, Widlund HR, Wucherpfennig KW, Puigserver P. Selective deficiency of mitochondrial respiratory complex I subunits Ndufs4/6 causes tumor immunogenicity. NATURE CANCER 2025; 6:323-337. [PMID: 39824999 DOI: 10.1038/s43018-024-00895-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/10/2024] [Indexed: 01/20/2025]
Abstract
Cancer cells frequently rewire their metabolism to support proliferation and evade immune surveillance, but little is known about metabolic targets that could increase immune surveillance. Here we show a specific means of mitochondrial respiratory complex I (CI) inhibition that improves tumor immunogenicity and sensitivity to immune checkpoint blockade (ICB). Targeted genetic deletion of either Ndufs4 or Ndufs6, but not other CI subunits, induces an immune-dependent growth attenuation in melanoma and breast cancer models. We show that deletion of Ndufs4 induces expression of the major histocompatibility complex (MHC) class I co-activator Nlrc5 and antigen presentation machinery components, most notably H2-K1. This induction of MHC-related genes is driven by a pyruvate dehydrogenase-dependent accumulation of mitochondrial acetyl-CoA, which leads to an increase in histone H3K27 acetylation within the Nlrc5 and H2-K1 promoters. Taken together, this work shows that selective CI inhibition restricts tumor growth and that specific targeting of Ndufs4 or Ndufs6 increases T cell surveillance and ICB responsiveness.
Collapse
Affiliation(s)
- Jiaxin Liang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tevis Vitale
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xixi Zhang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas D Jackson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deyang Yu
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
55
|
Bai X, Kang J, Wei S, Wang Y, Liu Y, Yuan B, Lu Q, Li H, Yan J, Yang X, Chang J. A pH responsive nanocomposite for combination sonodynamic-immunotherapy with ferroptosis and calcium ion overload via SLC7A11/ACSL4/LPCAT3 pathway. EXPLORATION (BEIJING, CHINA) 2025; 5:20240002. [PMID: 40040833 PMCID: PMC11875445 DOI: 10.1002/exp.20240002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/24/2024] [Indexed: 03/06/2025]
Abstract
Sonodynamic therapy offers a non-invasive approach to induce the death of tumor cells. By harnessing ultrasound waves in tandem with sonosensitizers, this method produces reactive oxygen species (ROS) that inflict oxidative damage upon tumor cells, subsequently causing their demise. Ferroptosis is a regulatory form of cell death that differs from other forms, characterized by iron accumulation, ROS accumulation, and lipid peroxidation. In the presented research, a nanoparticle formulation, parthenolide/ICG-CaCO3@lipid (PTL/ICG-CaCO3@Lip), has been engineered to amplify ferroptosis in tumor cells, positioning it as a potent agent for sonodynamic cancer immunotherapy. This nanoparticle significantly augments ROS levels within tumor cells, inducing oxidative stress that leads to cell death. The therapeutic potential of PTL/ICG-CaCO3@Lip, both in vivo and in vitro, has been convincingly demonstrated. Furthermore, RNA-seq analysis insights revealed that PTL/ICG-CaCO3@Lip facilitates tumor cell ferroptosis by regulating P53 to downregulate SLC7A11 protein expression, thereby inhibiting the glutamate-cystine antiporter system Xc- and stimulating ACSL4/LPCAT3 pathways. This pioneering work uncovers an innovative strategy for combatting tumors, leveraging enhanced oxidative stress to promote cell ferroptosis, and paves the way for groundbreaking cancer immunotherapeutic interventions.
Collapse
Affiliation(s)
- Xue Bai
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
| | - Jun Kang
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
- Department of Biological SciencesUniversity of Toronto ScarboroughTorontoCanada
| | | | - Yun Wang
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Yangsui Liu
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Bo Yuan
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Qian Lu
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
- Hepatopancreatobiliary CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
- School of Clinical MedicineTsinghua UniversityBeijingChina
| | - Huansong Li
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Jun Yan
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
- Hepatopancreatobiliary CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
- School of Clinical MedicineTsinghua UniversityBeijingChina
| | - Xi Yang
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
| | - Jin Chang
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
| |
Collapse
|
56
|
Guerrier L, Bacoeur-Ouzillou O, Touron J, Mezher S, Cassagnes L, Vieille-Marchiset A, Chanon S, Pereira B, Pezet D, Pinel A, Gagnière J, Malpuech-Brugère C, Richard R. Mitochondrial respiration in white adipose tissue is dependent on body mass index and tissue location in patients undergoing oncological or parietal digestive surgery. FASEB J 2025; 39:e70350. [PMID: 39856788 DOI: 10.1096/fj.202402243r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Adipose tissue (AT), is a major endocrine organ that plays a key role in health and disease. However, adipose dysfunctions, especially altered energy metabolism, have been under-investigated as white adipocytes have relatively low mitochondrial density. Nevertheless, recent studies suggest that mitochondria could play a major role in AT disorders and that AT mitochondrial activity could depend on adiposity level and location. This clinical study aimed to evaluate mitochondrial respiration and metabolism in human visceral (vAT) and subcutaneous (scAT) AT and their relationship with body mass index (BMI). This clinical study enrolled 67 patients (30 females/37 males) scheduled for digestive surgery without chemotherapy and parietal infection. BMI ranged from 15.4 to 51.9 kg·m-2 and body composition was estimated by computed tomographic images. Mitochondrial respiration was measured in situ in digitonin-permeabilized AT using high-resolution respirometry and a substrate/inhibitor titration approach. Protein levels of mitochondrial and lipid metabolism key elements were evaluated by Western blot. Maximal mitochondrial respiration correlated negatively with BMI (p < .01) and AT area (p < .001) regardless of the anatomical location. However, oxidative phosphorylation respiration was significantly higher in vAT (2.22 ± 0.15 pmol·sec-1·mg-1) than scAT (1.79 ± 0.17 pmol·sec-1·mg-1) (p < 0.001). In line with oxygraphy results, there were higher levels of mitochondrial respiratory chain complexes in low-BMI patients and vAT. Mitochondrial respiration decreased with increasing BMI in both scAT and vAT, without sex-associated difference. Mitochondrial respiration appeared to be higher in vAT than scAT. These differences were both qualitative and quantitative. Clinical Trials Registration IDNCT05417581.
Collapse
Affiliation(s)
- Lisa Guerrier
- INRAe, Human Nutrition Unit, Clermont Auvergne University, Clermont-Ferrand, France
| | - Ophélie Bacoeur-Ouzillou
- INRAe, Human Nutrition Unit, Clermont Auvergne University, Clermont-Ferrand, France
- Department of Digestive and Hepatobiliary Surgery-Liver Transplantation, CHU Estaing, Clermont-Ferrand, France
| | - Julianne Touron
- INRAe, Human Nutrition Unit, Clermont Auvergne University, Clermont-Ferrand, France
| | - Sami Mezher
- Department of Radiology, CHU Gabriel Montpied, Clermont-Ferrand, France
| | - Lucie Cassagnes
- Department of Radiology, CHU Gabriel Montpied, Clermont-Ferrand, France
| | | | - Stéphanie Chanon
- INRAe, INSERM, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France
| | - Bruno Pereira
- Biostatistics Unit, Clinical Research and Innovation Division, CHU Gabriel Montpied, Clermont-Ferrand, France
| | - Denis Pezet
- Department of Digestive and Hepatobiliary Surgery-Liver Transplantation, CHU Estaing, Clermont-Ferrand, France
| | - Alexandre Pinel
- INRAe, Human Nutrition Unit, Clermont Auvergne University, Clermont-Ferrand, France
| | - Johan Gagnière
- Department of Digestive and Hepatobiliary Surgery-Liver Transplantation, CHU Estaing, Clermont-Ferrand, France
| | | | - Ruddy Richard
- INRAe, Human Nutrition Unit, Clermont Auvergne University, Clermont-Ferrand, France
- Nutrition Exploration Unit, Human Nutrition Research Centre (CRNH) Auvergne, Clermont-Ferrand, France
- Department of Sport Medicine and Functional Explorations, CHU Gabriel Montpied, Clermont-Ferrand, France
| |
Collapse
|
57
|
Murillo Carrasco AG, Chammas R, Furuya TK. Mitochondrial DNA alterations in precision oncology: Emerging roles in diagnostics and therapeutics. Clinics (Sao Paulo) 2025; 80:100570. [PMID: 39884256 PMCID: PMC11830334 DOI: 10.1016/j.clinsp.2024.100570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 12/25/2024] [Indexed: 02/01/2025] Open
Abstract
Mitochondria are dynamic organelles essential for vital cellular functions, including ATP production, apoptosis regulation, and calcium homeostasis. Increasing research has highlighted the significance of mitochondrial DNA (mtDNA) content and alterations in the development and progression of various diseases, including cancer. The high mutation rate and vulnerability of mtDNA to damage make these alterations valuable biomarkers for cancer diagnosis, monitoring disease progression, detecting metastasis, and predicting treatment resistance across different tumor types. This review explores the emerging roles of mtDNA alterations in precision oncology, emphasizing their potential in theranostics. The authors explore the mechanisms by which mtDNA mutations contribute to tumorigenesis and therapy resistance, the impact of heteroplasmy in cancer biology, and the integration of mtDNA-based diagnostics with current therapeutic strategies. Additionally, the authors highlight the experimental tools and models currently used to investigate mtDNA alterations in cancer, including advanced sequencing technologies and animal models.
Collapse
Affiliation(s)
- Alexis Germán Murillo Carrasco
- Centro de Investigação Translacional em Oncologia (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil.
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil.
| | - Tatiane Katsue Furuya
- Centro de Investigação Translacional em Oncologia (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
58
|
Zhang T, Xu B. Didymin Inhibits Proliferation and Induces Apoptosis in Gastric Cancer Cells by Modulating the PI3K/Akt Pathway. Nutr Cancer 2025; 77:537-552. [PMID: 39849840 DOI: 10.1080/01635581.2025.2454050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Gastric cancer (GC) is a malignant tumor with high morbidity and mortality rates worldwide. This study aimed to investigate the effects and mechanisms of action of didymin, a dietary flavonoid glycoside, on GC treatment. Human GC cell lines Hs-746T and AGS were used to assess the effects of didymin on cell viability, cell proliferation, and cell cycle. The results showed that didymin decreased the proliferative capacity of GC cells and blocked cell cycle. Didymin decreased wound healing, invasion, and migration capacities of GC cells. Mitochondrial reactive oxygen species (ROS) levels and mitochondrial membrane potentials were reduced in cells treated with didymin. Network pharmacology analysis revealed that the therapeutic effects of didymin on AGS cells were related to the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. In vivo mouse xenograft studies confirmed that didymin treatment decreased tumor cell proliferation, cell cycle protein levels, and Akt phosphorylation. The present study demonstrated that didymin regulates mitochondrial function and the PI3K/Akt pathway to inhibit cell proliferation and induce apoptosis in GC cells in vitro and in vivo. Therefore, didymin is a promising drug for the treatment of GC.
Collapse
Affiliation(s)
- Tong Zhang
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Bin Xu
- Department of General Surgery, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
59
|
Akhunzianov AA, Rozhina EV, Filina YV, Rizvanov AA, Miftakhova RR. Resistance to Radiotherapy in Cancer. Diseases 2025; 13:22. [PMID: 39851486 PMCID: PMC11764699 DOI: 10.3390/diseases13010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Radiation therapy or radiotherapy is a medical treatment that uses high doses of ionizing radiation to eliminate cancer cells and shrink tumors. It works by targeting the DNA within the tumor cells restricting their proliferation. Radiotherapy has been used for treating cancer for more than 100 years. Along with surgery and chemotherapy, it is one of the three main and most common approaches used in cancer therapy. Nowadays, radiotherapy has become a standard treatment option for a wide range of cancers around the world, including lung, breast, cervical, and colorectal cancers. Around 50% of all patients will require radiotherapy, 60% of whom are treated with curative intent. Moreover, it is commonly used for palliative treatment. Radiotherapy provides 5-year local control and overall survival benefit in 10.4% and 2.4% of all cancer patients, respectively. The highest local control benefit is reported for cervical (33%), head and neck (32%), and prostate (26%) cancers. But no benefit is observed in pancreas, ovary, liver, kidney, and colon cancers. Such relatively low efficiency is related to the development of radiation resistance, which results in cancer recurrence, metastatic dissemination, and poor prognosis. The identification of radioresistance biomarkers allows for improving the treatment outcome. These biomarkers mainly include proteins involved in metabolism and cell signaling pathways.
Collapse
Affiliation(s)
- Almaz A. Akhunzianov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elvira V. Rozhina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yuliya V. Filina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| | - Regina R. Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
60
|
Deng L, Ran H, Yang D, Wang Z, Zhao P, Huang H, Wu Y, Zhang P. TOM40 as a prognostic oncogene for oral squamous cell carcinoma prognosis. BMC Cancer 2025; 25:92. [PMID: 39815211 PMCID: PMC11737269 DOI: 10.1186/s12885-024-13417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/31/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND To investigate the role of the translocase of the outer mitochondrial membrane 40 (TOM40) in oral squamous cell carcinoma (OSCC) with the aim of identifying new biomarkers or potential therapeutic targets. METHODS TOM40 expression level in OSCC was evaluated using datasets downloaded from The Cancer Genome Atlas (TCGA), as well as clinical data. The correlation between TOM40 expression level and the clinicopathological parameters and survival were analyzed in TCGA. The signaling pathways associated with TOM40 were identified through gene set enrichment analysis. A network of genes co-expressed with TOM40 was constructed and functionally annotated by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. The immune infiltration pattern in OSCC was analyzed in the TCGA-OSCC cohort using the CIBERSORT algorithm. Clinically significant factors of OSCC were screened through the expression levels of TOM40 and a clinically relevant nomogram was constructed. The TCGA-OSCC cohort was divided into the TOM40high and TOM40low groups and the correlation between TOM40 expression level and the sensitivity to frequently used chemotherapeutic drugs was evaluated. CCK-8 and colony formation assays were applied to determine the cell growth. RESULTS TOM40 was highly expressed in OSCC tissues and correlated negatively with the overall survival (P < 0.05). Patients with high TOM40 expression level showed worse prognosis. Furthermore, GO and KEGG enrichment analyses of the differentially expressed genes related to TOM40 showed that these genes are mainly associated with immunity and tumorigenesis. Immunological infiltration analysis has found that the expression levels of TOM40 are correlated with the proportions of several immune cells. Moreover, we found that TOM40 knockdown inhibited cell growth in OSCC cell lines. CONCLUSIONS Our results uncovered that TOM40 is a reliable prognostic marker and therapeutic target in OSCC.
Collapse
Affiliation(s)
- Lifei Deng
- Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, China
| | - Hong Ran
- Department of Otolaryngology-Head & Neck Surgery, Head and Neck Surgical Center, West China Hospital, Sichuan University, Chengdu, 610044, China
- Department of Otorhinolaryngology, Shenzhen Key Laboratory of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen Institute of Otorhinolaryngology, No. 3004 Longgang Avenue, Shenzhen, Guangdong, China
| | - Dunhui Yang
- Department of Otorhinolaryngology, Shenzhen Key Laboratory of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen Institute of Otorhinolaryngology, No. 3004 Longgang Avenue, Shenzhen, Guangdong, China
- Department of Graduate and Scientific Research, Zunyi Medical University, Zunyi, 563000, China
| | - Zhen Wang
- Department of Otorhinolaryngology, Shenzhen Key Laboratory of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen Institute of Otorhinolaryngology, No. 3004 Longgang Avenue, Shenzhen, Guangdong, China
| | - Peng Zhao
- Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, China
| | - Hengjie Huang
- School of Computer Science and Engineering, Yulin Normal University, No. 1303 Jiaoyu East Road, Yulin, 537000, Guangxi, China.
| | - Yongjin Wu
- Department of Otorhinolaryngology, Shenzhen Key Laboratory of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen Institute of Otorhinolaryngology, No. 3004 Longgang Avenue, Shenzhen, Guangdong, China.
| | - Peng Zhang
- Department of Otorhinolaryngology, Shenzhen Key Laboratory of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital, Shenzhen Institute of Otorhinolaryngology, No. 3004 Longgang Avenue, Shenzhen, Guangdong, China.
| |
Collapse
|
61
|
Doyle TM, Janes K, Xiao WH, Kolar GR, Luecke HF, Gratton MA, Tosh DK, Jacobson KA, Bennett GJ, Salvemini D. Mitochondrial A 3 Adenosine Receptor as a Mechanism for the Protective Effects of A 3AR Agonists on Chemotherapy-Induced Neuropathic Pain. J Neurosci 2025; 45:e1268242024. [PMID: 39653498 PMCID: PMC11735668 DOI: 10.1523/jneurosci.1268-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
Alterations in mitochondrial function are the linchpin in numerous disease states including in the development of chemotherapy-induced neuropathic pain (CIPN), a major dose-limiting toxicity of widely used chemotherapeutic cytotoxins. In CIPN, mitochondrial dysfunction is characterized by deficits in mitochondrial bioenergetics (e.g., decreased ATP production) that are thought to drive the degeneration of the peripheral nerve sensory axon terminal sensory arbors in the skin (the intraepidermal nerve fibers; IENFs) and induce abnormal spontaneous discharge in peripheral nerve sensory axons. Preserving mitochondrial function is anticipated to prevent CIPN. We have now discovered that the G-protein-coupled receptor, A3 adenosine receptor subtype (A3AR), is expressed on the mitochondrial outer membrane. Ex vivo application of a highly selective A3AR agonist, MRS5980, to saphenous nerve microfilaments harvested from male oxaliplatin-treated rats reversed the loss in ATP production underscoring mitoprotective effects resulting from A3AR activation on mitochondria. Moreover, in vivo administration of A3AR agonists to rats during oxaliplatin treatment was associated with reduced IENF loss and a lower incidence of spontaneous discharge in peripheral afferent axons. These effects are accompanied by improved mitochondrial ATP production in primary afferent sensory axons and overall inhibition of the development of neuropathic pain. These data identify for the first time mitochondrial A3AR and indicate that activation of A3AR protects mitochondrial function in primary afferent sensory axons against chemotherapy-induced neurotoxicity. Repurposing A3AR agonists that are already in clinical trials as anticancer agents as adjunct to chemotherapeutics will address a major unmet medical need for which there are no FDA-approved drugs.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| | - Kali Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Wen Hua Xiao
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Grant R Kolar
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Hans F Luecke
- Chemical Proteomics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael Anne Gratton
- Center for Sensory Neuroscience, Boys Town National Research Hospital, Boys Town, Nebraska 68010
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Gary J Bennett
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| |
Collapse
|
62
|
Huang A, Xue H, Xie T, Xiang L, Chen Z, Ma A, Yan H, Yuan J. A review of the pathogenesis of mitochondria in breast cancer and progress of targeting mitochondria for breast cancer treatment. J Transl Med 2025; 23:70. [PMID: 39815317 PMCID: PMC11734335 DOI: 10.1186/s12967-025-06077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025] Open
Abstract
With breast cancer being the most common tumor among women in the world today, it is also the leading cause of cancer-related deaths. Standard treatments include chemotherapy, surgery, endocrine therapy, and targeted therapy. However, the heterogeneity, drug resistance, and poor prognosis of breast cancer highlight an urgent need for further exploration of its underlying mechanisms. Mitochondria, highly dynamic intracellular organelles, play a pivotal role in maintaining cellular energy metabolism. Altered mitochondrial function plays a critical role in various diseases, and recent studies have elucidated its pathophysiological mechanisms in breast carcinogenesis. This review explores the role of mitochondrial dysfunction in breast cancer pathogenesis and assesses potential mitochondria-targeted therapies.
Collapse
Affiliation(s)
- Aoling Huang
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Haochen Xue
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Ting Xie
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Lingyan Xiang
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Zhengzhuo Chen
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Aolong Ma
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China.
| |
Collapse
|
63
|
Vijayanathan Y, Ho IAW. The Impact of Metabolic Rewiring in Glioblastoma: The Immune Landscape and Therapeutic Strategies. Int J Mol Sci 2025; 26:669. [PMID: 39859381 PMCID: PMC11765942 DOI: 10.3390/ijms26020669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by extensive metabolic reprogramming that drives tumor growth and therapeutic resistance. Key metabolic pathways, including glycolysis, lactate production, and lipid metabolism, are upregulated to sustain tumor survival in the hypoxic and nutrient-deprived tumor microenvironment (TME), while glutamine and tryptophan metabolism further contribute to the aggressive phenotype of GBM. These metabolic alterations impair immune cell function, leading to exhaustion and stress in CD8+ and CD4+ T cells while favoring immunosuppressive populations such as regulatory T cells (Tregs) and M2-like macrophages. Recent studies emphasize the role of slow-cycling GBM cells (SCCs), lipid-laden macrophages, and tumor-associated astrocytes (TAAs) in reshaping GBM's metabolic landscape and reinforcing immune evasion. Genetic mutations, including Isocitrate Dehydrogenase (IDH) mutations, Epidermal Growth Factor Receptor (EGFR) amplification, and Phosphotase and Tensin Homolog (PTEN) loss, further drive metabolic reprogramming and offer potential targets for therapy. Understanding the relationship between GBM metabolism and immune suppression is critical for overcoming therapeutic resistance. This review focuses on the role of metabolic rewiring in GBM, its impact on the immune microenvironment, and the potential of combining metabolic targeting with immunotherapy to improve clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Yuganthini Vijayanathan
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore 308433, Singapore;
| | - Ivy A. W. Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore 308433, Singapore;
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
64
|
Duan Z, Li L, Zhan Q, Chen J, Li Q, Liu R, Tu Y. Mitochondria-Targeting Type-I Photodynamic Therapy Based on Phenothiazine for Realizing Enhanced Immunogenic Cancer Cell Death via Mitochondrial Oxidative Stress. Int J Nanomedicine 2025; 20:125-139. [PMID: 39802375 PMCID: PMC11721160 DOI: 10.2147/ijn.s494970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Photo-immunotherapy faces challenges from poor immunogenicity and low response rate due to hypoxic microenvironment. This study presents Rh-PTZ, a small organic molecule with a D-π-A structure, that simultaneously amplifies mitochondria-targeted type-I PDT-dependent immune stimulation for the treatment of hypoxic cancer. Methods The hydrophobic Rh-PTZ was encapsulated into F127 to prepare Rh-PTZ nanoparticles (Rh-PTZ NPs). The type-I ROS generation ability, mitochondrial targeting capacity, and ICD triggering effect mediated by Rh-PTZ NPs under LED light irradiation were investigated. Based on a 4T1 subcutaneous tumor model, the in vivo biological safety assessment, in vivo NIR fluorescent imaging, and the efficacy of PDT were assessed. Results Rh-PTZ could efficiently accumulate in the mitochondrial site and induce O2 •- and •OH burst in situ under LED light irradiation, thereby causing severe mitochondrial dysfunction. Rh-PTZ can amplify mitochondrial stress-caused immunogenic cell death (ICD) to stimulate the immune response, promote the maturation of sufficient dendritic cells (DCs), enhance the infiltration of immune cells, and alleviate the tumor immunosuppressive microenvironment. Conclusion The mitochondria-targeting type-I PDT holds promise to enhance photo-immunotherapy for hypoxia tumor treatment and overcoming the limitations of traditional immunotherapy.
Collapse
Affiliation(s)
- Zeyu Duan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Lie Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qiyu Zhan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Jian Chen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qiyan Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Ruiyuan Liu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yinuo Tu
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| |
Collapse
|
65
|
Feijóo V, Tajada S, Méndez-Mena A, Núñez L, Villalobos C. Mitoception, or transfer of normal cell mitochondria to cancer cells, reverses remodeling of store-operated Ca 2+ entry in tumor cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119862. [PMID: 39437852 DOI: 10.1016/j.bbamcr.2024.119862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Most cancer cells show the Warburg effect, the rewiring of aerobic metabolism to glycolysis due to defective mitochondrial ATP synthesis. As a consequence, tumor cells display enhanced mitochondrial potential (∆Ψ), the driving force for mitochondrial Ca2+ uptake. Mitochondria control the Ca2+-dependent inactivation of store-operated channels (SOCs), leading to enhanced and sustained store-operated Ca2+ entry (SOCE) involved in cancer hallmarks. We asked here whether the transfer of mitochondria (mitoception) from normal cells to tumor cells may reverse SOCE remodeling in cancer cells. For this end, we labeled mitochondria in normal NCM460 human colonic cells, isolated them and transferred them to tumor HT29 cells. We tested the viability and efficiency of mitoception using flow cytometry and confocal microscopy, as well as calcium imaging to investigate the effects of mitoception on SOCE. Our results show that mitoception of tumor HT29 cells with normal mitochondria restores a low ∆Ψ and SOCE. Conversely, self-mitoception of tumor HT29 cells with tumor cell mitochondria increases further ∆Ψ and SOCE, thus excluding the possibility that effects of mitoception are due to increased mitochondrial mass. Strikingly, mitoception of normal NCM460 cells with tumor cell mitochondria has no effects on either ∆Ψ or SOCE. These results are consistent with the previous proposal that transformed mitochondria may modulate SOC channels involved in SOCE. Further research is warranted to test whether mitoception of cancer cells with normal mitochondria may reverse Ca2+ remodeling associated to cancer.
Collapse
Affiliation(s)
- Verónica Feijóo
- Excellence Unit Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Sendoa Tajada
- Excellence Unit Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain; Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Alejandra Méndez-Mena
- Excellence Unit Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Lucía Núñez
- Excellence Unit Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain; Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Carlos Villalobos
- Excellence Unit Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain.
| |
Collapse
|
66
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
67
|
Zhao X, Zhang J, Li C, Kuang W, Deng J, Tan X, Li C, Li S. Mitochondrial mechanisms in Treg cell regulation: Implications for immunotherapy and disease treatment. Mitochondrion 2025; 80:101975. [PMID: 39491776 DOI: 10.1016/j.mito.2024.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining immune homeostasis and preventing autoimmune diseases. Recent advances in immunometabolism have revealed the pivotal role of mitochondrial dynamics and metabolism in shaping Treg functionality. Tregs depend on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to support their suppressive functions and long-term survival. Mitochondrial processes such as fusion and fission significantly influence Treg activity, with mitochondrial fusion enhancing bioenergetic efficiency and reducing reactive oxygen species (ROS) production, thereby promoting Treg stability. In contrast, excessive mitochondrial fission disrupts ATP synthesis and elevates ROS levels, impairing Treg suppressive capacity. Furthermore, mitochondrial ROS act as critical signaling molecules in Treg regulation, where controlled levels stabilize FoxP3 expression, but excessive ROS leads to mitochondrial dysfunction and immune dysregulation. Mitophagy, as part of mitochondrial quality control, also plays an essential role in preserving Treg function. Understanding the intricate interplay between mitochondrial dynamics and Treg metabolism provides valuable insights for developing novel therapeutic strategies to treat autoimmune disorders and enhance immunotherapy in cancer.
Collapse
Affiliation(s)
- Xiaozhen Zhao
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Junmei Zhang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Caifeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Weiying Kuang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jianghong Deng
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xiaohua Tan
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chao Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shipeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
68
|
Shen Y, Zhang T, Jia X, Xi F, Jing W, Wang Y, Huang M, Na R, Xu L, Ji W, Qiao Y, Zhang X, Sun W, Li S, Wu J. MEF2A, a gene associated with mitochondrial biogenesis, promotes drug resistance in gastric cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167497. [PMID: 39237047 DOI: 10.1016/j.bbadis.2024.167497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Chemotherapeutic resistance is a major obstacle to the effectiveness of cisplatin-based chemotherapy for gastric cancer (GC), leading to treatment failure and poor survival rates. However, the underlying mechanisms are not fully understood. Our study demonstrated that the transcription factor myocyte enhancer factor 2A (MEF2A) plays a role in chemotherapeutic drug resistance by regulating the transcription of PGC1α and KEAP1, promoting mitochondrial biogenesis. It was found that increased MEF2A expression is linked with poor prognosis, cisplatin insensitivity, and mitochondrial function in GC. MEF2A overexpression significantly decreases GC cell sensitivity in vitro and in vivo, while MEF2A knockdown enhances the sensitivity to cisplatin. Mechanistically, MEF2A activates the transcription of PGC1α, leading to increased mitochondrial biogenesis. In addition, MEF2A inhibits KEAP1 transcription, reduces NRF2 ubiquitination degradation, and activates the KEAP1/NRF2 signaling pathway, which modulates the reactive oxygen species level. The present study identifies MEF2A as a new critical oncogene involved in GC chemoresistance, suggesting a novel therapeutic target for GC.
Collapse
Affiliation(s)
- Yao Shen
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Tong Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xueyuan Jia
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Fei Xi
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wanting Jing
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yusi Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Min Huang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lidan Xu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wei Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yuandong Qiao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xuelong Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shuijie Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China; Future Medical Laboratory, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
69
|
Gonçalves FA, da Silva Bittencourt L, Barbosa S, Diel LF, Bernardi L, Matte C, Lamers ML. Energy Metabolic Profile in Oral Potentially Malignant Disorders and Oral Squamous Cell Carcinoma: A Preliminary Landscape of Warburg Effect in Oral Cancer. Mol Carcinog 2025; 64:126-137. [PMID: 39412414 DOI: 10.1002/mc.23831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 12/13/2024]
Abstract
We hypothesized that cell energy metabolic profiles correlate with normal, dysplastic, and tumor cell/tissue statuses and may be indicators of aggressiveness in oral squamous cell carcinoma (OSCC) cells. The energy-related proteins that were differentially expressed in human OSCC fragments (n = 3) and their adjacent epithelial tissue (TAE) were verified using mass spectrometry (MS). Immunohistochemistry for 4-hydroxynonenal (4-HNE) was performed to evaluate the oxidative stress patterns in OSCC (n = 10), epithelial dysplasia (n = 9), and normal epithelial (n = 4) biopsies. The metabolic energy profile of OSCC aggressiveness was investigated in human OSCC cell lines with different levels of epithelial-mesenchymal transition proteins. The genes associated with the proteins found by MS in this study were analyzed using survival analysis (OS), whereas the genes associated with a poorer prognosis were analyzed using context-specific expression, Gene Ontology (GO) and Cancer Hallmarks for function enrichment analysis. The rationale for all experimental approach was to investigate whether the variation in energy metabolism profile accompanies the different phenotypes (from epithelial to mesenchymal) during the epithelial-mesenchymal transition. All OSCC fragments exhibited an increase in glycolysis-related proteins and a decrease in mitochondrial activity compared to the TAE region (p < 0.05), probably due to the downregulation of pyruvate dehydrogenase and antioxidant proteins. Additionally, the OSCC cell lines with a mesenchymal profile (SCC4, SCC9, and SCC25) had a lower mitochondrial mass and membrane potential and generated lower levels of reactive oxygen and nitrogen species than the TAE region. When we analyzed 4-HNE, the reactive species levels were increased in the epithelial regions of OSCC and potentially malignant lesions. A decrease in the levels of 4-HNE/reactive species was observed in the connective tissue underlying the dysplastic regions and the OSCC invasion zone. Based on this scenario, aggressive OSCC is associated with high glycolytic and oxidative metabolism and low mitochondrial and antioxidant activities, which vary according to the differentiation level of the tumor cells and the stage of carcinogenesis.
Collapse
Affiliation(s)
- Francisca Aurina Gonçalves
- Basic Research Center in Dentistry, School of Dentistry, Federal University of Rio Grande of Sul, Porto Alegre, Brazil
| | - Leonardo da Silva Bittencourt
- Basic Research Center in Dentistry, School of Dentistry, Federal University of Rio Grande of Sul, Porto Alegre, Brazil
- Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- State Department of Education of Rio Grande do Sul-State Professional School of Health, Clinical Hospital of Porto Alegre (EPS-HCPA), Porto Alegre, Brazil
| | - Silvia Barbosa
- Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Leonardo Francisco Diel
- Basic Research Center in Dentistry, School of Dentistry, Federal University of Rio Grande of Sul, Porto Alegre, Brazil
| | - Lisiane Bernardi
- Basic Research Center in Dentistry, School of Dentistry, Federal University of Rio Grande of Sul, Porto Alegre, Brazil
- Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiane Matte
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcelo Lazzaron Lamers
- Basic Research Center in Dentistry, School of Dentistry, Federal University of Rio Grande of Sul, Porto Alegre, Brazil
- Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
70
|
Zhu S, Chen C, Wang M, Liu Y, Li B, Qi X, Song M, Liu X, Feng J, Liu J. Pan-cancer association of a mitochondrial function score with genomic alterations and clinical outcome. Sci Rep 2024; 14:31430. [PMID: 39733076 PMCID: PMC11682264 DOI: 10.1038/s41598-024-83022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Mitochondria are pivotal in cellular energy metabolism and have garnered significant attention for their roles in cancer progression and therapy resistance. Despite this, the functional diversity of mitochondria across various cancer types remains inadequately characterized. This study seeks to fill this knowledge gap by introducing and validating MitoScore-a novel metric designed to quantitatively assess mitochondrial function across a wide array of cancers. Our investigation evaluates the capacity of MitoScore not only to distinguish between tumor and adjacent normal tissues but also to serve as a predictive marker for clinical outcomes. We analyzed gene expression data from 24 cancer types and corresponding normal tissues using the TCGA database. MitoScore was calculated by summing the normalized expression levels of six mitochondrial genes known to be consistently altered across multiple cancers. Differential gene expression was assessed using DESeq2, with a focus on identifying significant changes in mitochondrial function. MitoScore's associations with tumor proliferation, hypoxia, aneuploidy, and clinical outcomes were evaluated using Spearman's correlation, linear regression, and Kaplan-Meier survival analyses. MitoScore was significantly higher in tumor tissues compared to normal tissues across most cancer types (p < 0.001). It positively correlated with tumor proliferation rates (r = 0.46), hypoxia scores (r = 0.61), and aneuploidy (r = 0.44), indicating its potential as a marker of aggressive tumor behavior. High MitoScore was also associated with poorer prognosis in several cancer types, suggesting its utility as a predictive biomarker for clinical outcomes. This study introduces MitoScore, a metric for mitochondrial activity often elevated in tumors and linked to poor prognosis. It correlates positively with hypoxia and negatively with stromal and immune infiltration, highlighting mitochondria's role in the tumor microenvironment. MitoScore's association with genomic instability, such as aneuploidy, suggests mitochondrial dysfunction contributes to cancer progression. Despite challenges in mitochondrial-targeted therapies, MitoScore may identify tumors responsive to such treatments, warranting further research for clinical application.
Collapse
Affiliation(s)
- Shikun Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Chen Chen
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Min Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Yue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xing Qi
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
- Ziyang People's Hospital, Ziyang, Sichaun, China
| | - Miao Song
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xuexue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| |
Collapse
|
71
|
Huang Y, Wu G, Bi G, Cheng L, Liang J, Li M, Zhang H, Shan G, Hu Z, Chen Z, Lin Z, Jiang W, Wang Q, Xi J, Yin S, Zhan C. Unveiling chemotherapy-induced immune landscape remodeling and metabolic reprogramming in lung adenocarcinoma by scRNA-sequencing. eLife 2024; 13:RP95988. [PMID: 39729352 DOI: 10.7554/elife.95988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
Chemotherapy is widely used to treat lung adenocarcinoma (LUAD) patients comprehensively. Considering the limitations of chemotherapy due to drug resistance and other issues, it is crucial to explore the impact of chemotherapy and immunotherapy on these aspects. In this study, tumor samples from nine LUAD patients, of which four only received surgery and five received neoadjuvant chemotherapy, were subjected to scRNA-seq analysis. In vitro and in vivo assays, including flow cytometry, immunofluorescence, Seahorse assay, and tumor xenograft models, were carried out to validate our findings. A total of 83,622 cells were enrolled for subsequent analyses. The composition of cell types exhibited high heterogeneity across different groups. Functional enrichment analysis revealed that chemotherapy drove significant metabolic reprogramming in tumor cells and macrophages. We identified two subtypes of macrophages: Anti-mac cells (CD45+CD11b+CD86+) and Pro-mac cells (CD45+CD11b+ARG +) and sorted them by flow cytometry. The proportion of Pro-mac cells in LUAD tissues increased significantly after neoadjuvant chemotherapy. Pro-mac cells promote tumor growth and angiogenesis and also suppress tumor immunity. Moreover, by analyzing the remodeling of T and B cells induced by neoadjuvant therapy, we noted that chemotherapy ignited a relatively more robust immune cytotoxic response toward tumor cells. Our study demonstrates that chemotherapy induces metabolic reprogramming within the tumor microenvironment of LUAD, particularly affecting the function and composition of immune cells such as macrophages and T cells. We believe our findings will offer insight into the mechanisms of drug resistance and provide novel therapeutic targets for LUAD in the future.
Collapse
Affiliation(s)
- Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Cheng
- Department of Pathology, Albert Einstein College of Medicine, Bronx, United States
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Zhang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Sichuan, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junjie Xi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shanye Yin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, United States
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
72
|
Wang Y, Hu W, Zhou B, Zhao Y, Tang Y, Deng Z, Chen M. Mitochondrial transcription elongation factor TEFM promotes malignant progression of gliomas. Cancer Cell Int 2024; 24:429. [PMID: 39719635 DOI: 10.1186/s12935-024-03617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Gliomas are the most common tumors of the central nervous system, with glioblastoma (GBM) being particularly aggressive and fatal. Current treatments for GBM, including surgery and chemotherapy, are limited by tumor aggressiveness and the blood-brain barrier. Therefore, understanding the molecular mechanisms driving GBM growth is essential. Mitochondria, key players in cellular energy production, have been implicated in cancer development. In this study, we investigated the expression of mitochondrial transcription elongation factor (TEFM) in gliomas and its potential role in tumor progression. Analysis of data from The Cancer Genome Atlas (TCGA) revealed that TEFM transcript levels were significantly higher in glioma tissues compared to adjacent normal tissues. High TEFM expression was associated with poor survival outcomes in glioma patients. Furthermore, TEFM was notably upregulated in glioma tissue and in primary glioma cells derived from local patients, while its expression was relatively low in normal tissues and astrocytes. Silencing or knockout of TEFM significantly inhibited glioma cell growth, proliferation, clonogenicity, migration, and invasion, while inducing apoptosis and activating caspases. In contrast, ectopic overexpression of TEFM promoted tumorigenic activity, enhancing the malignant behavior of glioma cells. Co-expression analysis identified a strong correlation between TEFM and the epithelial-mesenchymal transition (EMT) pathway in gliomas. Notably, the expression of EMT markers, such as N-cadherin and Vimentin, decreased upon TEFM knockdown or knockout. Additionally, TEFM depletion impaired mitochondrial function, disrupting the mitochondrial respiratory chain in glioma cells. In vivo experiments demonstrated that TEFM knockout effectively suppressed the growth of subcutaneous glioma xenografts in nude mice. Collectively, these findings highlight the critical role of TEFM in GBM growth and invasion, suggesting that it could serve as a promising therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yin Wang
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China
| | - Wenxuan Hu
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Boya Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yu Zhao
- Department of Soochow Medical College, Soochow University, Suzhou, China
| | - Yufei Tang
- Department of Soochow Medical College, Soochow University, Suzhou, China
| | - Zhiyong Deng
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China.
| | - Minbin Chen
- Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital, Soochow University, Suzhou, China.
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
73
|
Shao N, Yang Y, Hu G, Luo Q, Cheng N, Chen J, Huang Y, Zhang H, Luo L, Xiao Z. Synergistic enhancement of low-dose radiation therapy via cuproptosis and metabolic reprogramming for radiosensitization in in situ hepatocellular carcinoma. J Nanobiotechnology 2024; 22:772. [PMID: 39696547 DOI: 10.1186/s12951-024-03011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is a primary clinical approach for cancer treatment, but its efficacy is often hindered by various challenges, especially radiation resistance, which greatly compromises the therapeutic effectiveness of RT. Mitochondria, central to cellular energy metabolism and regulation of cell death, play a critical role in mechanisms of radioresistance. In this context, cuproptosis, a novel copper-induced mitochondria-respiratory-dependent cell death pathway, offers a promising avenue for radiosensitization. RESULTS In this study, an innovative theranostic nanoplatform was designed to induce cuproptosis in synergy with low-dose radiation therapy (LDRT, i.e., 0.5-2 Gy) for the treatment of in situ hepatocellular carcinoma (HCC). This approach aims to reverse the hypoxic tumor microenvironment, promoting a shift in cellular metabolism from glycolysis to oxidative phosphorylation (OXPHOS), thereby enhancing sensitivity to cuproptosis. Concurrently, the Fenton-like reaction ensures a sustained supply of copper and depletion of glutathione (GSH), inducing cuproptosis, disrupting mitochondrial function, and interrupting the energy supply. This strategy effectively overcomes radioresistance and enhances the therapeutic efficacy against tumors. CONCLUSIONS In conclusion, this study elucidates the intricate interactions among tumor hypoxia reversal, cuproptosis, metabolic reprogramming, and radiosensitization, particularly in the context of treating in situ hepatocellular carcinoma, thereby providing a novel paradigm for radiotherapy.
Collapse
Affiliation(s)
- Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yongqing Yang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Qiao Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Nianlan Cheng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Hong Zhang
- Department of Interventional Radiology and Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523067, China.
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- Department of Radiology and Nuclear Medicine, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan, 517000, China.
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
74
|
Qin Y, Gao Y, Wu D, Liu QQ, Su C, Liu G, Yang L, Zhao MG, Yao JY. Ailanthone blocks mitophagy to promote mtDNA leakage through BAX-BAK1 pores and suppress hepatocellular carcinoma cell proliferation. Front Pharmacol 2024; 15:1509482. [PMID: 39723259 PMCID: PMC11668963 DOI: 10.3389/fphar.2024.1509482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC), the third leading cancer mortality worldwide, shows rising incidence. The mitochondria in HCC cells are prone to damage from metabolic stress and oxidative stress, necessitating heightened mitophagy for mitochondrial homeostasis and cell survival. Thus, mitophagy inhibition is a promising HCC therapy. The traditional Chinese medicinal herb ailanthone have proved promote mitochondrial dysfunction and inhibits HCC. However, the underlying mechanism remains unclear. Methods CCK8 assay was applied to detect the proliferation. JC-1, MitoTracker Red/Green and MitoSOX staining were applied to detect the mitochondrial homeostasis. Inflammatory factors were analysed via ELISA and WB assay. Mitochondria and cytoplasm separation, genome extraction and qPCR were used to detect mitochondrial DNA (mtDNA) leakage. Mitochondria ultrastructure was detected by transmission electron microscopy. WB and IHC experiments were applied to detect protein expression. Protein-protein interactions detected by immunoprecipitation and immunofluorescence imaging. The in vivo antitumor effect was validated by the xenograft mouse model. Results In this study, we demonstrated the potent anti-HCC properties of ailanthone and revealed its molecular mechanism. In vitro studies demonstrated that ailanthone effectively inhibited PINK1-PRKN mediated mitophagy and promoted BAX-BAK1 mitochondrial pores formation through PRKN inhibition. This process led to the mitochondrial mtDNA leakage into the cytoplasm, which subsequently triggered the induction of inflammatory factors. The inhibition of mitophagy and the activation of inflammatory response ultimately led to HCC proliferation inhibition. In vivo studies demonstrated that ailanthone exhibited stronger anti-HCC activity than 5-Fluorouracil (5-FU), with no significant adverse effects on animal body weight or the physiological functions of vital organs. Conclusion This study highlighted the efficacy of ailanthone against HCC and elucidated its underlying molecular mechanisms, suggesting the promising therapeutic potential of ailanthone for HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Le Yang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Ming-Gao Zhao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Jing-Yue Yao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
75
|
Huo G, Lin Y, Liu L, He Y, Qu Y, Liu Y, Zhu R, Wang B, Gong Q, Han Z, Yin H. Decoding ferroptosis: transforming orthopedic disease management. Front Pharmacol 2024; 15:1509172. [PMID: 39712490 PMCID: PMC11659002 DOI: 10.3389/fphar.2024.1509172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
As a mechanism of cell death, ferroptosis has gained popularity since 2012. The process is distinguished by iron toxicity and phospholipid accumulation, in contrast to autophagy, apoptosis, and other cell death mechanisms. It is implicated in the advancement of multiple diseases across the body. Researchers currently know that osteosarcoma, osteoporosis, and other orthopedic disorders are caused by NRF2, GPX4, and other ferroptosis star proteins. The effective relief of osteoarthritis symptoms from deterioration has been confirmed by clinical treatment with multiple ferroptosis inhibitors. At the same time, it should be reminded that the mechanisms involved in ferroptosis that regulate orthopedic diseases are not currently understood. In this manuscript, we present the discovery process of ferroptosis, the mechanisms involved in ferroptosis, and the role of ferroptosis in a variety of orthopedic diseases. We expect that this manuscript can provide a new perspective on clinical diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guanlin Huo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Yi Qu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Bo Wang
- Department of Orthopaedics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Qing Gong
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhongyu Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongbing Yin
- Orthopedic Center, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
76
|
Du Y, Wang L, Perez-Castro L, Conacci-Sorrell M, Sieber M. Non-cell autonomous regulation of cell-cell signaling and differentiation by mitochondrial ROS. J Cell Biol 2024; 223:e202401084. [PMID: 39535785 PMCID: PMC11561560 DOI: 10.1083/jcb.202401084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondrial reactive oxygen species (ROS) function intrinsically within cells to induce cell damage, regulate transcription, and cause genome instability. However, we know little about how mitochondrial ROS production non-cell autonomously impacts cell-cell signaling. Here, we show that mitochondrial dysfunction inhibits the plasma membrane localization of cell surface receptors that drive cell-cell communication during oogenesis. Within minutes, we found that mitochondrial ROS impairs exocyst membrane binding and leads to defective endosomal recycling. This endosomal defect impairs the trafficking of receptors, such as the Notch ligand Delta, during oogenesis. Remarkably, we found that overexpressing RAB11 restores ligand trafficking and rescues the developmental defects caused by ROS production. ROS production from adjacent cells acutely initiates a transcriptional response associated with growth and migration by suppressing Notch signaling and inducing extra cellualr matrix (ECM) remodeling. Our work reveals a conserved rapid response to ROS production that links mitochondrial dysfunction to the non-cell autonomous regulation of cell-cell signaling.
Collapse
Affiliation(s)
- Yipeng Du
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
77
|
Chen F, Xue Y, Zhang W, Zhou H, Zhou Z, Chen T, YinWang E, Li H, Ye Z, Gao J, Wang S. The role of mitochondria in tumor metastasis and advances in mitochondria-targeted cancer therapy. Cancer Metastasis Rev 2024; 43:1419-1443. [PMID: 39307891 PMCID: PMC11554835 DOI: 10.1007/s10555-024-10211-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Mitochondria are central actors in diverse physiological phenomena ranging from energy metabolism to stress signaling and immune modulation. Accumulating scientific evidence points to the critical involvement of specific mitochondrial-associated events, including mitochondrial quality control, intercellular mitochondrial transfer, and mitochondrial genetics, in potentiating the metastatic cascade of neoplastic cells. Furthermore, numerous recent studies have consistently emphasized the highly significant role mitochondria play in coordinating the regulation of tumor-infiltrating immune cells and immunotherapeutic interventions. This review provides a comprehensive and rigorous scholarly investigation of this subject matter, exploring the intricate mechanisms by which mitochondria contribute to tumor metastasis and examining the progress of mitochondria-targeted cancer therapies.
Collapse
Affiliation(s)
- Fanglu Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiyi Zhou
- The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy YinWang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
78
|
Hu C, Zheng Z, Pang S, Zhu Y, Jie J, Lai Z, Zeng X, Xiao Y, Chen Z, Zhao J, Du Y, Li F, Wang Q, Tan W. Chimeric SFT2D2-TBX19 Promotes Prostate Cancer Progression by Encoding TBX19-202 Protein and Stabilizing Mitochondrial ATP Synthase through ATP5F1A Phosphorylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408426. [PMID: 39540264 DOI: 10.1002/advs.202408426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Specific chimeric RNAs and their products are consistently regarded as ideal tumor diagnostic markers and therapeutic targets. Chimeric RNAs can mediate tumor cell plasticity, neuroendocrine processes, polarization of tumor-associated macrophages, and resistance to chemotherapy and immunotherapy. However, the discovery of chimeric RNAs in prostate cancer is still in its early stages. This study identifies the chimeric SFT2D2-TBX19 as a novel transcript encoding the TBX19-202 protein. Both TBX19-202 and its parental TBX19, which share homologous amino acid sequences, enhance prostate cancer cell proliferation, migration, and invasion. Additionally, SFT2D2-TBX19 also functions as a lncRNA, interacting with the ATP synthase F1 subunit ATP5F1A, thereby increasing ATP5F1A phosphorylation mediated by TNK2/ACK1, which stabilizes the interaction between ATP5F1A and ATP5F1B. The region spanning 1801-2400 bp of SFT2D2-TBX19 and the intermediate structural domain of ATP5F1A are crucial functional areas. This stabilization of ATP5F1A and ATP5F1B enhances mitochondrial ATP synthase activity and ATP production. Even under conditions of mitochondrial vulnerability, SFT2D2-TBX19 protects mitochondrial structural stability to maintain prostate cancer cell proliferation. This research provides comprehensive evidence that chimeric SFT2D2-TBX19 promotes prostate cancer progression by encoding the TBX19-202 protein and stabilizing mitochondrial ATP synthase via ATP5F1A phosphorylation. These findings highlight SFT2D2-TBX19 as a potential therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Chenxi Hu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Zaosong Zheng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Shiyu Pang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yuanchao Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jirong Jie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhuocheng Lai
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xiangbo Zeng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yongyuan Xiao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhifeng Chen
- Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jingjing Zhao
- Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yuejun Du
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
79
|
Farahani N, Alimohammadi M, Raei M, Nabavi N, Aref AR, Hushmandi K, Daneshi S, Razzaghi A, Taheriazam A, Hashemi M. Exploring the dual role of endoplasmic reticulum stress in urological cancers: Implications for tumor progression and cell death interactions. J Cell Commun Signal 2024; 18:e12054. [PMID: 39691874 PMCID: PMC11647052 DOI: 10.1002/ccs3.12054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 12/19/2024] Open
Abstract
The endoplasmic reticulum (ER) is crucial for maintaining calcium balance, lipid biosynthesis, and protein folding. Disruptions in ER homeostasis, often due to the accumulation of misfolded or unfolded proteins, lead to ER stress, which plays a significant role in various diseases, especially cancer. Urological cancers, which account for high male mortality worldwide, pose a persistent challenge due to their incurability and tendency to develop drug resistance. Among the numerous dysregulated biological mechanisms, ER stress is a key factor in the progression and treatment response of these cancers. This review highlights the dual role of aberrant ER stress activation in urologic cancers, affecting both tumor growth and therapeutic outcomes. While ER stress can support tumor growth through pro-survival autophagy, it primarily inhibits cancer progression via apoptosis and pro-death autophagy. Interestingly, ER stress can paradoxically aid cancer progression through mechanisms such as exosome-mediated immune evasion. Additionally, the review examines how pharmacological interventions, particularly with phytochemicals, can stimulate ER stress-mediated tumor suppression. Key regulators, including PERK, IRE1α, and ATF6, are discussed for their roles in upregulating CHOP levels and triggering apoptosis. In conclusion, a deeper understanding of ER stress in urological cancers not only clarifies the complex interactions between cellular stress and cancer progression but also provides new opportunities for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mina Alimohammadi
- Department of ImmunologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Mehdi Raei
- Health Research CenterLife Style InstituteBaqiyatallah University of Medical SciencesTehranIran
| | | | - Amir Reza Aref
- Department of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kiavash Hushmandi
- Nephrology and Urology Research CenterClinical Sciences InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Salman Daneshi
- Department of Public HealthSchool of HealthJiroft University of Medical SciencesJiroftIran
| | - Alireza Razzaghi
- Social Determinants of Health Research CenterResearch Institute for Prevention of Non‐Communicable DiseasesQazvin University of Medical SciencesQazvinIran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of OrthopedicsFaculty of MedicineTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| |
Collapse
|
80
|
Kong W, Meng Q, Kong RM, Zhao Y, Qu F. Valence-Transforming O 2-Depleting Nano-Assembly Enable In Situ Tumor Depositional Embolization. Adv Healthc Mater 2024; 13:e2402899. [PMID: 39328009 DOI: 10.1002/adhm.202402899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Indexed: 09/28/2024]
Abstract
Abnormal metabolism and blood supply/O2 imbalance in tumor cells affect drug transport delivery and increase the difficulty of tumor treatment. Controlling tumor growth by inhibiting tumor cell metabolism and regulating progressive embolization in the tumor region provides an innovative basis for constructing tumor therapeutic models. A highly biocompatible and efficient O2-depleting agent has been investigated to enable in situ precipitation and embolization within the tumor microenvironment. In situ deformation embolizer, Fe-GA@CaCO3 nano-assembly (GA: gallic acid), can convert into the large granular size embolization components of Fe(III) precipitates and affluent Ca2+ within the tumor microenvironment. In situ progressive O2 depletion produces Fe(III) precipitates that embolize tumor regions, isolating O2 and nutrients by blocking supply. Meanwhile, affluent Ca2+ acts on the intracellular, causing mitochondrial dysfunction through calcium overload and contributing to irreversible tumor cell damage. Both internal and external routes work synergistically to produce precise functional inhibition of tumors from the inside out, simultaneously responding to both intracellular and the corresponding tumor regions, providing an innovative solution for anti-tumor therapy.
Collapse
Affiliation(s)
- Weiheng Kong
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, China
| | - Qingyao Meng
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, China
| | - Rong-Mei Kong
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, China
| | - Yan Zhao
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong, 273165, China
| | - Fengli Qu
- Department of Pathology, Cancer Hospital of Zhejiang Province, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| |
Collapse
|
81
|
Fan Q, Xiao K, A R, Gao LP, Wu YZ, Chen DD, Hu C, Jia XX, Liu CM, Liu X, Chen C, Shi Q, Dong XP. Accumulation of Prion Triggers the Enhanced Glycolysis via Activation of AMKP Pathway in Prion-Infected Rodent and Cell Models. Mol Neurobiol 2024; 61:9810-9834. [PMID: 37726499 DOI: 10.1007/s12035-023-03621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023]
Abstract
Mitochondrial dysfunction is one of the hallmarks in the pathophysiology of prion disease and other neurodegenerative diseases. Various metabolic dysfunctions are identified and considered to contribute to the progression of some types of neurodegenerative diseases. In this study, we evaluated the status of glycolysis pathway in prion-infected rodent and cell models. The levels of the key enzymes, hexokinase (HK), phosphofructokinase (PFK), and pyruvate kinase (PK) were significantly increased, accompanying with markedly downregulated mitochondrial complexes. Double-stained IFAs revealed that the increased HK2 and PFK distributed widely in GFAP-, Iba1-, and NeuN-positive cells. We also identified increased levels of AMP-activated protein kinase (AMPK) and the downstream signaling. Changes of AMPK activity in prion-infected cells by the AMPK-specific inhibitor or activator induced the corresponding alterations not only in the downstream signaling, but also the expressions of three key kinases in glycolysis pathway and the mitochondrial complexes. Transient removal or complete clearance of prion propagation in the prion-infected cells partially but significantly reversed the increases of the key enzymes in glycolysis, the upregulation of AMPK signaling pathway, and the decreases of the mitochondrial complexes. Measurements of the cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) showed lower OCR and higher ECAR in prion-infected cell line, which were sufficiently reversed by clearance of prion propagation. Those data indicate a metabolic reprogramming from oxidative phosphorylation to glycolysis in the brains during the progression of prion disease. Accumulation of PrPSc is critical for the switch to glycolysis, largely via activating AMPK pathway.
Collapse
Affiliation(s)
- Qin Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruhan A
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li-Ping Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yue-Zhang Wu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong-Dong Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chao Hu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Xi Jia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chu-Mou Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qi Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiao-Ping Dong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
82
|
Kamble OS, Chatterjee R, Abishek KG, Chandra J, Alsayari A, Wahab S, Sahebkar A, Kesharwani P, Dandela R. Small molecules targeting mitochondria as an innovative approach to cancer therapy. Cell Signal 2024; 124:111396. [PMID: 39251050 DOI: 10.1016/j.cellsig.2024.111396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cellular death evasion is a defining characteristic of human malignancies and a significant contributor to therapeutic inefficacy. As a result of oncogenic inhibition of cell death mechanisms, established therapeutic regimens seems to be ineffective. Mitochondria serve as the cellular powerhouses, but they also function as repositories of self-destructive weaponry. Changes in the structure and activities of mitochondria have been consistently documented in cancer cells. In recent years, there has been an increasing focus on using mitochondria as a targeted approach for treating cancer. Considerable attention has been devoted to the development of delivery systems that selectively aim to deliver small molecules called "mitocans" to mitochondria, with the ultimate goal of modulating the physiology of cancer cells. This review summarizes the rationale and mechanism of mitochondrial targeting with small molecules in the treatment of cancer, and their impact on the mitochondria. This paper provides a concise overview of the reasoning and mechanism behind directing treatment towards mitochondria in cancer therapy, with a particular focus on targeting using small molecules. This review also examines diverse small molecule types within each category as potential therapeutic agents for cancer.
Collapse
Affiliation(s)
- Omkar S Kamble
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - K G Abishek
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
83
|
Wang L, Tian S, Deng S, Wu J, Wang H, Guo X, Han C, Ren W, Han Y, Zhou J, Lin Y, Bu M. Design and synthesis of novel mitochondria-targeted ergosterol peroxide derivatives as potential anti-cancer agents. Bioorg Chem 2024; 153:107862. [PMID: 39362080 DOI: 10.1016/j.bioorg.2024.107862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Ergosterol peroxide (EP) is a natural steroid compound that has been reported to have significant antitumor activity. However, its poor water solubility and cellular uptake mean that it has weak efficacy against tumor cells. Herein, we designed and synthesized a series of EP derivatives with mitochondrial targeting properties. Of these, compound 15a showed an IC50 value of 0.32 μM against MCF-7 cells, which was 67-fold higher than that of the parental EP (IC50 = 21.46 μM), and was better than cisplatin (IC50 = 4.23 μM), had a selectivity index of 25.28 (IC50MCF-10A/IC50MCF-7). Additionally, compound 15a promoted an increase in intracellular reactive oxygen species levels and a decrease in mitochondrial membrane potential, and blocked the cell cycle in the G0/G1 phase. In a mouse model of breast cancer, 15a showed 89.85 % tumor inhibition at a dose of 20 mg/kg, which is similar to the therapeutic effect of the cisplatin. On the basis of these results, 15a could be considered for further preclinical evaluation for cancer therapy.
Collapse
Affiliation(s)
- Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Shuang Tian
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jiale Wu
- College of Pharmacy, Hainan University, Haikou 570228, Hainan, PR China
| | - Haijun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Xiaoshan Guo
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Cuicui Han
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Wenkang Ren
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yinglong Han
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jianwen Zhou
- Research Institute of Medicine & Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| |
Collapse
|
84
|
Alberto PL, Ashim S, Megan K, Wei Z, Nestor G, Matthew Z, Dayana M, Marcelo V, John W. Salivary Gland Oncocytomas. A Systematic Review. Head Neck Pathol 2024; 18:126. [PMID: 39614964 PMCID: PMC11608208 DOI: 10.1007/s12105-024-01730-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/01/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Oncocytoma is a primary benign epithelial neoplasm comprising less than 2% of salivary tumors with a low recurrence rate. METHODS A systematic review of documented case reports and case series of oncocytomas is presented. Searches from different databases were performed to identify articles from 1956 to 2024. The variables included were gender, age, symptoms, duration time before diagnosis, type of gland, histological features, special or immunohistochemical evaluation, treatment, follow-up, recurrence, and relation with a medical condition or syndrome. RESULTS Of the 147 cases reported, 53.1% affected females, and 46.9% were in males. The average age was 58.7 years, and the mean size was 2.3 cm. The most common clinical presentation was swelling (92.6%) and 66.7% were asymptomatic. The parotid was the most commonly affected gland with 66% of cases, the submandibular gland with 23.3%, and the minor salivary glands with Phosphotungstic acid-hematoxylin (PTAH) was the most common special stain used in 36.7%, followed by a combination with Periodic acid-Schiff (PAS) with and without diastase in 26.6%. Excisional biopsy was the most common treatment in 38.1% followed by superficial parotidectomy in 32.7%. Follow-up was 34.7 months on average. Bilateral oncocytomas were found in 4.8% with a 6 to 1 female-male proportion. Recurrence was found in 2.7% and association with Birt-Hogg-Dube (BHD) syndrome was 8.2%. CONCLUSION Salivary oncocytoma is a rare epithelial neoplasm with nonspecific clinical presentations. Diagnosis can be suspected on cytology and confirmed by histologic examination. The lesion has an indolent clinical course and most of the reported cases did not recur. There seems to be an association between bilateral oncocytomas and females and a low but interesting association with BHD. Overall, this review serves to better highlight the features of this rare benign neoplasm.
Collapse
Affiliation(s)
| | | | - Koch Megan
- Baylor University Medical Center, Dallas, USA
| | - Zhang Wei
- Baylor University Medical Center, Dallas, USA
| | | | | | | | | | - Wright John
- Texas A&M University School of Dentistry, Dallas, USA
| |
Collapse
|
85
|
Caporali S, Butera A, Ruzza A, Zampieri C, Bantula' M, Scharsich S, Ückert AK, Celardo I, Kouzel IU, Leanza L, Gruber A, Montero J, D'Alessandro A, Brunner T, Leist M, Amelio I. Selective metabolic regulations by p53 mutant variants in pancreatic cancer. J Exp Clin Cancer Res 2024; 43:310. [PMID: 39587609 PMCID: PMC11590503 DOI: 10.1186/s13046-024-03232-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Approximately half of all human cancers harbour mutations in the p53 gene, leading to the generation of neomorphic p53 mutant proteins. These mutants can exert gain-of-function (GOF) effects, potentially promoting tumour progression. However, the clinical significance of p53 GOF mutations, as well as the selectivity of individual variants, remains controversial and unclear. METHODS To elucidate the metabolic regulations and molecular underpinnings associated with the specific p53R270H and p53R172H mutant variants (the mouse equivalents of human p53R273H and p53R175H, respectively), we employed a comprehensive approach. This included integrating global metabolomic analysis with epigenomic and transcriptomic profiling in mouse pancreatic cancer cells. Additionally, we assessed metabolic parameters such as oxygen consumption rate and conducted analyses of proliferation and cell-cell competition to validate the biological impact of metabolic changes on pancreatic ductal adenocarcinoma (PDAC) phenotype. Our findings were further corroborated through analysis of clinical datasets from human cancer cohorts. RESULTS Our investigation revealed that the p53R270H variant, but not p53R172H, sustains mitochondrial function and energy production while also influencing cellular antioxidant capacity. Conversely, p53R172H, while not affecting mitochondrial metabolism, attenuates the activation of pro-tumorigenic metabolic pathways such as the urea cycle. Thus, the two variants selectively control different metabolic pathways in pancreatic cancer cells. Mechanistically, p53R270H induces alterations in the expression of genes associated with oxidative stress and reduction in mitochondrial respiration. In contrast, p53R172H specifically impacts the expression levels of enzymes involved in the urea metabolism. However, our analysis of cell proliferation and cell competition suggested that the expression of either p53R270H or p53R172H does not influence confer any selective advantage to this cellular model in vitro. Furthermore, assessment of mitochondrial priming indicated that the p53R270H-driven mitochondrial effect does not alter cytochrome c release or the apoptotic propensity of pancreatic cancer cells. CONCLUSIONS Our study elucidates the mutant-specific impact of p53R270H and p53R172H on metabolism of PDAC cancer cells, highlighting the need to shift from viewing p53 mutant variants as a homogeneous group of entities to a systematic assessment of each specific p53 mutant protein. Moreover, our finding underscores the importance of further exploring the significance of p53 mutant proteins using models that more accurately reflect tumor ecology.
Collapse
Affiliation(s)
- Sabrina Caporali
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alessio Butera
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alessia Ruzza
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Carlotta Zampieri
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, San Diego, CA, USA
| | - Marina Bantula'
- Department of Biomedical Sciences, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain
| | - Sandra Scharsich
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Anna-Katerina Ückert
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ivana Celardo
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ian U Kouzel
- Applied Bioinformatic Group, University of Konstanz, Constance, Germany
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy
| | - Andreas Gruber
- Applied Bioinformatic Group, University of Konstanz, Constance, Germany
| | - Joan Montero
- Department of Biomedical Sciences, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
| | | | - Thomas Brunner
- Chair for in Biochemical Pharmacology, University of Konstanz, Constance, Germany
| | - Marcel Leist
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ivano Amelio
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
86
|
Wang Y, Liu C, Ma X, Filppula A, Cui Y, Ye J, Zhang H. Encapsulated mitochondria to reprogram the metabolism of M2-type macrophages for anti-tumor therapy. NANOSCALE 2024; 16:20925-20939. [PMID: 39469868 DOI: 10.1039/d4nr02471k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
M2-type macrophages (M2Φ) play a pro-tumorigenic role and are closely associated with tumor development, where metabolic dysregulation exacerbates the immunosuppressive tumor microenvironment and fosters tumor growth. Mitochondria serve as the regulatory center of cellular metabolism, yet effective methods to modulate M2Φ mitochondria within the tumor microenvironment remain lacking. In this study, we developed a technique utilizing the bio-encapsulation of mitochondria in Zeolitic Imidazolate Framework-8 (ZiF-8), referred to as Mito@ZiF-8. Our findings demonstrated that this coating protects intact mitochondria and preserves their bioactivity over an extended period after isolation. We successfully delivered Mito@ZiF-8 into M2Φ, which inhibited the secretion of pro-inflammatory factors, promoted the release of anti-inflammatory factors, and reprogrammed M2Φ metabolism. This innovative approach has the potential to reduce breast cancer cell metastasis and enhance sensitivity to chemotherapy drugs such as 6-thioguanine, cisplatin, and doxorubicin (Dox). Mito@ZiF-8 aims to reprogram the M2Φ microenvironment to support anti-tumor therapies, offering a novel strategy for improving the effectiveness of breast cancer treatment.
Collapse
Affiliation(s)
- Yonghui Wang
- Department of Thoracic Surgery, Organ Transplantation Center, The First Hospital of Jilin University, Changchun, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku 20520, Finland.
| | - Chang Liu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku 20520, Finland.
| | - Xiaodong Ma
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku 20520, Finland.
| | - Anne Filppula
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku 20520, Finland.
| | - Youbin Cui
- Department of Thoracic Surgery, Organ Transplantation Center, The First Hospital of Jilin University, Changchun, China
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Hongbo Zhang
- Department of Thoracic Surgery, Organ Transplantation Center, The First Hospital of Jilin University, Changchun, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku 20520, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| |
Collapse
|
87
|
Luo Y, Liu R, Zhang H, Wang H, Yin H, Tian G, Wang B, Yan Y, Ding Z, Dai J, Niu L, Yuan G, Pan Y. Amantadine against glioma via ROS-mediated apoptosis and autophagy arrest. Cell Death Dis 2024; 15:834. [PMID: 39548081 PMCID: PMC11568115 DOI: 10.1038/s41419-024-07228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Glioma is a common primary nervous system malignant tumor with poor overall cure rate and low survival rate, yet successful treatment still remains a challenge. Here, we demonstrated that amantadine (AMT) exhibits the powerful anti-glioma effect by promoting apoptosis and autophagy in vivo and in vitro. Mechanistically, amantadine induces a large amount of reactive oxygen species (ROS) accumulation in glioma cells, and then triggers apoptosis by destroying mitochondria. In addition, amantadine induces the initiation of autophagy and inhibits the fusion of autophagosome and lysosome, consequently performing an anti-glioma role. Taken together, our findings suggest that amantadine could be a promising anti-glioma drug that inhibits glioma cells by inducing apoptosis and autophagy, which may provide a novel potential treatment option for patients.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - He Zhang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hongyu Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hang Yin
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guopeng Tian
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bo Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yunji Yan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zilin Ding
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Junqiang Dai
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Liang Niu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
88
|
Fang K, Xu H, Yuan S, Li X, Chen X, Fan X, Gao X, Zhang L, Sun S, Zhu X. LncRNA mediated metabolic reprogramming: the chief culprits of solid tumor malignant progression: an update review. Nutr Metab (Lond) 2024; 21:89. [PMID: 39516895 PMCID: PMC11549785 DOI: 10.1186/s12986-024-00866-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Metabolism reprogramming (MR) is one of the top ten hallmarks of malignant tumors. The aberrant activation of MR has been recognized as a critical contributory factor to the malignant progression of solid tumors. Moreover, various long non-coding RNAs (lncRNAs) are implicated in the aberrant activation of MR in solid tumor cells. Therefore, in this review, we mainly focus on summarizing the functional relevance and molecular mechanistic underpinnings of lncRNAs in modulating MR of solid tumors by targeting glucose metabolism, lipid metabolism, affecting mitochondrial function, and regulating interactions between tumor and non-tumor cells in tumor microenvironment. Besides, we also underscore the potential for constructing lncRNAs-centered tumor metabolic regulation networks and developing novel anti-tumor strategies by targeting lncRNAs and abnormal MR. Ultimately, this review seeks to offer new targets and avenues for the clinical treatment of solid tumors in the future.
Collapse
Affiliation(s)
- Kun Fang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Huizhe Xu
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiaoyu Chen
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiushi Fan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiaoxin Gao
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Lu Zhang
- Department of Human Resources, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China.
| | - Shulan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China.
| | - Xudong Zhu
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning, China.
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
89
|
Nevoit G, Jarusevicius G, Potyazhenko M, Mintser O, Bumblyte IA, Vainoras A. Mitochondrial Dysfunction and Risk Factors for Noncommunicable Diseases: From Basic Concepts to Future Prospective. Diseases 2024; 12:277. [PMID: 39589951 PMCID: PMC11592525 DOI: 10.3390/diseases12110277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Noncommunicable diseases (NCDs) are a very important medical problem. The key role of mitochondrial dysfunction (MD) in the occurrence and progression of NCDs has been proven. However, the etiology and pathogenesis of MD itself in many NCDs has not yet been clarified, which makes it one of the most serious medical problems in the modern world, according to many scientists. METHODS An extensive research in the literature was implemented in order to elucidate the role of MD and NCDs' risk factors in the pathogenesis of NCDs. RESULTS The authors propose to take a broader look at the problem of the pathogenesis of NCDs. It is important to understand exactly how NCD risk factors lead to MD. The review is structured in such a way as to answer this question. Based on a systematic analysis of scientific data, a theoretical concept of modern views on the occurrence of MD under the influence of risk factors for the occurrence of NCDs is presented. This was done in order to update MD issues in clinical medicine. MD and NCDs progress throughout a patient's life. Based on this, the review raised the question of the existence of an NCDs continuum. CONCLUSIONS MD is a universal mechanism that causes organ dysfunction and comorbidity of NCDs. Prevention of MD involves diagnosing and eliminating the factors that cause it. Mitochondria are an important therapeutic target.
Collapse
Affiliation(s)
- Ganna Nevoit
- Laboratory of Population Studies, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Gediminas Jarusevicius
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Maksim Potyazhenko
- Department of Internal Medicine and Emergency Medicine, Poltava State Medical University, 36011 Poltava, Ukraine;
| | - Ozar Mintser
- Department of Fundamental Disciplines and Informatics, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine;
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Alfonsas Vainoras
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| |
Collapse
|
90
|
Wu Y, Jia C, Liu W, Zhan W, Chen Y, Lu J, Bao Y, Wang S, Yu C, Zheng L, Sun L, Song Z. Sodium citrate targeting Ca 2+/CAMKK2 pathway exhibits anti-tumor activity through inducing apoptosis and ferroptosis in ovarian cancer. J Adv Res 2024; 65:89-104. [PMID: 38724006 PMCID: PMC11518946 DOI: 10.1016/j.jare.2024.04.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 10/21/2024] Open
Abstract
INTRODUCTION Ovarian cancer (OC) is known for its high mortality rate. Although sodium citrate has anti-tumor effects in various cancers, its effect and mechanism in OC remain unclear. OBJECTIVES To analyze the inhibitory effect of sodium citrate on ovarian cancer cells and the underlying mechanism. METHODS Cell apoptosis was examined by TUNEL staining, flow cytometry, and ferroptosis was examined intracellular Fe2+, MDA, LPO assays, respectively. Cell metabolism was examined by OCR and ECAR measurements. Immunoblotting and immunoprecipitation were used to elucidate the mechanism. RESULTS This study suggested that sodium citrate not only promoted ovarian cancer cell apoptosis but also triggeredferroptosis, manifested as elevated levels of Fe2+, LPO, MDA andlipid ROS production. On one hand, sodium citrate treatment led to a decrease of Ca2+ content in the cytosol by chelatingCa2+, which further inhibited the Ca2+/CAMKK2/AKT/mTOR signaling, thereby suppressing HIF1α-dependent glycolysis pathway and inducing cell apoptosis. On the other hand, the chelation of Ca2+ by sodium citrate resulted in inactivation of CAMKK2 and AMPK, leading to increase of NCOA4-mediated ferritinophagy, causing increased intracellular Fe2+ levels. More importantly, the inhibition of Ca2+/CAMKK2/AMPK signaling pathway reduced the activity of the MCU and Ca2+ concentration within the mitochondria, resulting in an increase in mitochondrial ROS. Additionally, metabolomic analysis indicated that sodium citrate treatment significantly increased de novo lipid synthesis. Altogether, these factors contributed to ferroptosis. As expected, Ca2+ supplementation successfully reversed the cell death and decreased tumor growth induced by sodium citrate. Inspiringly, it was found that coadministration of sodium citrate increased the sensitivity of OC cells to chemo-drugs. CONCLUSION These results revealed that the sodium citrate exerted its anti-cancer activity by inhibiting Ca2+/CAMKK2-dependent cell apoptosis and ferroptosis. Sodium citrate will hopefully serve as a prospective compound for OC treatment and for improvingthe efficacy of chemo-drugs.
Collapse
Affiliation(s)
- Yulun Wu
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China
| | - Chaoran Jia
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China; National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Wei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Wei Zhan
- The Hospital of Northeast Normal University, Changchun 130024, China
| | - Yao Chen
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China; National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Junlin Lu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Yongli Bao
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China
| | - Shuyue Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Chunlei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Lihua Zheng
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China.
| | - Zhenbo Song
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine Products, Northeast Normal University, Changchun 130024, China.
| |
Collapse
|
91
|
Burvenich IJG, Osellame LD, Rigopoulos A, Huynh N, Cao Z, Hoogenraad NJ, Scott AM. Targeting Fn14 as a therapeutic target for cachexia reprograms the glycolytic pathway in tumour and brain in mice. Eur J Nucl Med Mol Imaging 2024; 51:3854-3862. [PMID: 39060375 PMCID: PMC11527931 DOI: 10.1007/s00259-024-06836-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE Cachexia is a complex syndrome characterized by unintentional weight loss, progressive muscle wasting and loss of appetite. Anti-Fn14 antibody (mAb 002) targets the TWEAK receptor (Fn14) in murine models of cancer cachexia and can extend the lifespan of mice by restoring the body weight of mice. Here, we investigated glucose metabolic changes in murine models of cachexia via [18F]FDG PET imaging, to explore whether Fn14 plays a role in the metabolic changes that occur during cancer cachexia. METHODS [18F]FDG PET/MRI imaging was performed in cachexia-inducing tumour models versus models that do not induce cachexia. SUVaverage was calculated for all tumours via volume of interest (VOI) analysis of PET/MRI overlay images using PMOD software. RESULTS [18F]FDG PET imaging demonstrated increased tumour and brain uptake in cachectic versus non-cachectic tumour-bearing mice. Therapy with mAb 002 was able to reduce [18F]FDG uptake in tumours (P < 0.05, n = 3). Fn14 KO tumours did not induce body weight loss and did not show an increase in [18F]FDG tumour and brain uptake over time. In non-cachectic mice bearing Fn14 KO tumours, [18F]FDG tumour uptake was significantly lower (P < 0.01) than in cachectic mice bearing Fn14 WT counterparts. As a by-product of glucose metabolism, l-lactate production was also increased in cachexia-inducing tumours expressing Fn14. CONCLUSION Our results demonstrate that Fn14 receptor activation is linked to glucose metabolism of cachexia-inducing tumours.
Collapse
Affiliation(s)
- Ingrid Julienne Georgette Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Laura Danielle Osellame
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3083, Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia
| | - Nhi Huynh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Zhipeng Cao
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3083, Australia
| | - Nicholas Johannes Hoogenraad
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3083, Australia
| | - Andrew Mark Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Melbourne, Heidelberg, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3083, Australia.
- Department of Medicine, University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
92
|
Fan S, Guo J, Nie H, Xiong H, Xia Y. Aberrant Energy Metabolism in Tumors and Potential Therapeutic Targets. Genes Chromosomes Cancer 2024; 63:e70008. [PMID: 39584783 PMCID: PMC11587691 DOI: 10.1002/gcc.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Energy metabolic reprogramming is frequently observed during tumor progression as tumor cells necessitate adequate energy production for rapid proliferation. Although current medical research shows promising prospects in studying the characteristics of tumor energy metabolism and developing anti-tumor drugs targeting energy metabolism, there is a lack of systematic compendiums and comprehensive reviews in this field. The objective of this study is to conduct a systematic review on the characteristics of tumor cells' energy metabolism, with a specific focus on comparing abnormalities between tumor and normal cells, as well as summarizing potential targets for tumor therapy. Additionally, this review also elucidates the aberrant mechanisms underlying four major energy metabolic pathways (glucose, lipid, glutamine, and mitochondria-dependent) during carcinogenesis and tumor progression. Through the utilization of graphical representations, we have identified anomalies in crucial energy metabolism pathways, encompassing transporter proteins (glucose transporter, CD36, and ASCT2), signaling molecules (Ras, AMPK, and PTEN), as well as transcription factors (Myc, HIF-1α, CREB-1, and p53). The key molecules responsible for aberrant energy metabolism in tumors may serve as potential targets for cancer therapy. Therefore, this review provides an overview of the distinct energy-generating pathways within tumor cells, laying the groundwork for developing innovative strategies for precise cancer treatment.
Collapse
Affiliation(s)
- Shuhao Fan
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Jianhua Guo
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Hui Nie
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJiningShandongPeople's Republic of China
| | - Yong Xia
- Shandong First Medical UniversityJinanShandongPeople's Republic of China
- College of Medical EngineeringJining Medical UniversityJiningShandongPeople's Republic of China
| |
Collapse
|
93
|
Martinez-Bernabe T, Pons DG, Oliver J, Sastre-Serra J. Oxidative Phosphorylation as a Predictive Biomarker of Oxaliplatin Response in Colorectal Cancer. Biomolecules 2024; 14:1359. [PMID: 39595536 PMCID: PMC11591675 DOI: 10.3390/biom14111359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Oxaliplatin is successfully used on advanced colorectal cancer to eradicate micro-metastasis, whereas its benefits in the early stages of colorectal cancer remains controversial since approximately 30% of patients experience unexpected relapses. Herein, we evaluate the efficacy of oxidative phosphorylation as a predictive biomarker of oxaliplatin response in colorectal cancer. We found that non-responding patients exhibit low oxidative phosphorylation activity, suggesting a poor prognosis. To reach this conclusion, we analyzed patient samples of individuals treated with oxaliplatin from the GSE83129 dataset, and a set of datasets validated using ROCplotter, selecting them based on their response to the drug. By analyzing multiple oxaliplatin-resistant and -sensitive cell lines, we identified oxidative phosphorylation KEGG pathways as a valuable predictive biomarker of oxaliplatin response with a high area under the curve (AUC = 0.843). Additionally, some oxidative phosphorylation-related biomarkers were validated in primary- and metastatic-derived tumorspheres, confirming the results obtained in silico. The low expression of these biomarkers is clinically relevant, indicating poor prognosis with decreased overall and relapse-free survival. This study proposes using oxidative phosphorylation-related protein expression levels as a predictor of responses to oxaliplatin-based treatments to prevent relapse and enable a more personalized therapy approach. Our results underscore the value of oxidative phosphorylation as a reliable marker for predicting the response to oxaliplatin treatment in colorectal cancer.
Collapse
Affiliation(s)
- Toni Martinez-Bernabe
- Gruop Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (D.G.P.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Daniel G. Pons
- Gruop Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (D.G.P.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Jordi Oliver
- Gruop Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (D.G.P.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Jorge Sastre-Serra
- Gruop Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (D.G.P.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
94
|
ZHANG Z, LIU H, CHEN J. [Role of Mitochondria in Ferroptosis and Its Relationship to Tumors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:785-791. [PMID: 39631835 PMCID: PMC11629089 DOI: 10.3779/j.issn.1009-3419.2024.102.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Indexed: 12/07/2024]
Abstract
Ferroptosis is a recently discovered form of cell death that is distinct from apoptosis, characterized primarily by the accumulation of intracellular iron and increased levels of lipid peroxidation. Resistance of tumor cells to ferroptosis can promote tumorigenesis and tumor progression. Various compounds can influence tumor development by triggering ferroptosis. Ferroptosis involves complex regulatory mechanisms, with mitochondria serving as both an iron storage and metabolic center, playing a crucial regulatory role in ferroptosis. This review discusses ferroptosis and its three stages and the role of ferroptosis in tumorigenesis, progression, and treatment, as well as the regulatory mechanisms of mitochondria in ferroptosis.
.
Collapse
|
95
|
Chiu CF, Guerrero JJG, Regalado RRH, Zhou J, Notarte KI, Lu YW, Encarnacion PC, Carles CDD, Octavo EM, Limbaroc DCI, Saengboonmee C, Huang SY. Insights into Metabolic Reprogramming in Tumor Evolution and Therapy. Cancers (Basel) 2024; 16:3513. [PMID: 39456607 PMCID: PMC11506062 DOI: 10.3390/cancers16203513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer remains a global health challenge, characterized not just by uncontrolled cell proliferation but also by the complex metabolic reprogramming that underlies its development and progression. Objectives: This review delves into the intricate relationship between cancer and its metabolic alterations, drawing an innovative comparison with the cosmological concepts of dark matter and dark energy to highlight the pivotal yet often overlooked role of metabolic reprogramming in tumor evolution. Methods: It scrutinizes the Warburg effect and other metabolic adaptations, such as shifts in lipid synthesis, amino acid turnover, and mitochondrial function, driven by mutations in key regulatory genes. Results: This review emphasizes the significance of targeting these metabolic pathways for therapeutic intervention, outlining the potential to disrupt cancer's energy supply and signaling mechanisms. It calls for an interdisciplinary research approach to fully understand and exploit the intricacies of cancer metabolism, pointing toward metabolic reprogramming as a promising frontier for developing more effective cancer treatments. Conclusion: By equating cancer's metabolic complexity with the enigmatic nature of dark matter and energy, this review underscores the critical need for innovative strategies in oncology, highlighting the importance of unveiling and targeting the "dark energy" within cancer cells to revolutionize future therapy and research.
Collapse
Affiliation(s)
- Ching-Feng Chiu
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- Taipei Medical University Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Jonathan Jaime G. Guerrero
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Ric Ryan H. Regalado
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City 1101, Philippines;
| | - Jiayan Zhou
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Yu-Wei Lu
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
| | - Paolo C. Encarnacion
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan; (J.J.G.G.); (Y.-W.L.); (P.C.E.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
- Department of Industrial Engineering and Management, Yuan Ze University, 135 Yuan-Tung Road, Chung-Li 32003, Taiwan
| | - Cidne Danielle D. Carles
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Edrian M. Octavo
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
| | - Dan Christopher I. Limbaroc
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines; (C.D.D.C.); (E.M.O.); (D.C.I.L.)
- College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
96
|
Jiang C, Wu W, Jiang X, Qian K. Integrative multi-omics analysis unveils the connection between transcriptomic characteristics associated with mitochondria and the tumor immune microenvironment in lower-grade gliomas. Sci Rep 2024; 14:23675. [PMID: 39390013 PMCID: PMC11467307 DOI: 10.1038/s41598-024-74281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Lower-grade gliomas (LGGs) exhibit diverse clinical behaviors and varying immune infiltration levels. Mitochondria have been implicated in numerous cancer pathogenesis and development, including LGGs. However, the precise biological functions of mitochondrial genes in shaping the immune landscape and the prognostic significance of LGGs remain elusive. Utilizing the Mito-Carta3.0 database, we curated a total of 1136 genes implicated in mitochondrial functions. By leveraging the expression profiles of 1136 genes related to mitochondria, we successfully categorized LGGs into four distinctive mitochondria-related transcriptome (MRT) subtypes. Our thorough analysis conclusively demonstrated that these subtypes exhibited marked disparities. To enable a personalized and integrated evaluation of LGG patients, we developed a prognostic signature known as MRT-related prognostic signature (MTRS). MTRS demonstrated correlation with mitochondria-related transcriptome (MRT) subtypes, allowing the assessment of patients' prognosis and immune microenvironment. We conducted a detailed exploration of the single-cell distribution of MTRS in lower-grade gliomas and verified the core genes of MTRS within the spatial transcriptome of these tumors. Furthermore, our study pinpointed MGME1 as the pivotal gene in the model, functioning as an oncogene that exerts influence on cell proliferation and migration capabilities. Our research highlights the importance of mitochondrial transcriptomic features in LGGs, offering paths for tailored therapies.
Collapse
Affiliation(s)
- Cheng Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Kang Qian
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
97
|
Li L, Liu C, Wang R, Yang X, Wei X, Chu C, Zhang G, Liu C, Cui W, Xu H, Wang K, An L, Li X. A novel role for WZ3146 in the inhibition of cell proliferation via ERK and AKT pathway in the rare EGFR G719X mutant cells. Sci Rep 2024; 14:22895. [PMID: 39358400 PMCID: PMC11447065 DOI: 10.1038/s41598-024-73293-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Mutations in the epidermal growth factor receptor (EGFR) gene are common driver oncogenes in non-small cell lung cancer (NSCLC). Studies have shown that afatinib is beneficial for NSCLC patients with rare EGFR mutations. However, the effectiveness of tyrosine kinase inhibitors (TKIs) against the G719X (G719A, G719C and G719S) mutation has not been fully established. Herein, using the CRISPR method, the EGFR G719X mutant cell lines were constructed to assess the sensitivity of the rare mutation G719X in NSCLC. WZ3146, a novel mutation-selective EGFR inhibitor, was conducted transcriptome sequencing and in vitro experiments. The results showed that WZ3146 induced cytotoxic effects, inhibited growth vitality and proliferation via ERK and AKT pathway in the EGFR G719X mutant cells. Our findings suggest that WZ3146 may be a promising treatment option for NSCLC patients with the EGFR exon 18 substitution mutation G719X.
Collapse
Affiliation(s)
- Lanxin Li
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
- Institute of Metabolism and Health, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Chenyang Liu
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Rui Wang
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Xiaolin Yang
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Xiangkai Wei
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Chunhong Chu
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China
| | - Guoliang Zhang
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China
| | - Chenxue Liu
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Wenrui Cui
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Huixia Xu
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Ke Wang
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China
| | - Lei An
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China.
- Institutes of Traditional Chinese Medicine, School of Pharmacy, Henan University, Kaifeng, 475000, Henan, China.
| | - Xiaodong Li
- No.115, Ximen Avenue, Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, 475000, China.
| |
Collapse
|
98
|
Zhao B, Liang Z, Zhang L, Jiang L, Xu Y, Zhang Y, Zhang R, Wang C, Liu Z. Ponicidin Promotes Hepatocellular Carcinoma Mitochondrial Apoptosis by Stabilizing Keap1-PGAM5 Complex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406080. [PMID: 39116422 PMCID: PMC11481384 DOI: 10.1002/advs.202406080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Ponicidin is a diterpenoid with demonstrated antitumor activity in clinical trials. However, the specific function and mechanism of action against hepatocellular carcinoma (HCC) remain unknown. In this study, it is found that ponicidin significantly inhibited the proliferation and migration of HCC cells. It is shown that ponicidin targets Keap1 and promotes the formation of the Keap1-PGAM5 complex, leading to the ubiquitination of PGAM5, using biotin-labeled ponicidin for target fishing and the HuProtTM Human Proteome Microarray V4.0. Ponicidin is found to activate the cysteine-dependent mitochondrial pathway via PGAM5, resulting in mitochondrial damage and ROS production, thereby promoting mitochondrial apoptosis in HepG2 cells. The first in vitro cocrystal structure of the PGAM5 IE 12-mer peptide and the Keap1 Kelch domain is obtained. Using molecular dynamics simulations to confirm the binding of ponicidin to the Keap1-PGAM5 complex. Based on the depth-based dynamic simulation, it is found that ponicidin can induce the tightening of the Keap1-PGAM5 interaction pocket, thereby stabilizing the formation of the protein complex. Finally, it is observed that ponicidin effectively inhibited tumor growth and promoted tumor cell apoptosis in a BALB/c nude mouse xenograft tumor model. The results provide insight into the anti-HCC properties of ponicidin based on a mechanism involving the Keap1-PGAM5 complex.
Collapse
Affiliation(s)
- Bixin Zhao
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Lisheng Zhang
- Research Center of Integrative MedicineSchool of Basic Medical ScienceGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Lin Jiang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Ying Zhang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| |
Collapse
|
99
|
Zhang W, Tang Y, Yang P, Chen Y, Xu Z, Qi C, Huang H, Liu R, Qin H, Ke H, Huang C, Xu F, Pang P, Zhao Z, Shan H, Xiao F. TMX2 potentiates cell viability of hepatocellular carcinoma by promoting autophagy and mitophagy. Autophagy 2024; 20:2146-2163. [PMID: 38797513 PMCID: PMC11423672 DOI: 10.1080/15548627.2024.2358732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
The dysregulation of membrane protein expression has been implicated in tumorigenesis and progression, including hepatocellular carcinoma (HCC). In this study, we aimed to identify membrane proteins that modulate HCC viability. To achieve this, we performed a CRISPR activation screen targeting human genes encoding membrane-associated proteins, revealing TMX2 as a potential driver of HCC cell viability. Gain- and loss-of-function experiments demonstrated that TMX2 promoted growth and tumorigenesis of HCC. Clinically, TMX2 was an independent prognostic factor for HCC patients. It was significantly upregulated in HCC tissues and associated with poor prognosis of HCC patients. Mechanistically, TMX2 was demonstrated to promote macroautophagy/autophagy by facilitating KPNB1 nuclear export and TFEB nuclear import. In addition, TMX2 interacted with VDAC2 and VADC3, assisting in the recruitment of PRKN to defective mitochondria to promote cytoprotective mitophagy during oxidative stress. Most interestingly, HCC cells responded to oxidative stress by upregulating TMX2 expression and cell autophagy. Knockdown of TMX2 enhanced the anti-tumor effect of lenvatinib. In conclusion, our findings emphasize the pivotal role of TMX2 in driving the HCC cell viability by promoting both autophagy and mitophagy. These results suggest that TMX2 May serve as a prognostic marker and promising therapeutic target for HCC treatment.Abbreviation: CCCP: Carbonyl cyanide 3-chlorophenylhydrazone; Co-IP: co-immunoprecipitation; CRISPR: clustered regularly interspaced short palindromic repeat; ER: endoplasmic reticulum; HCC: hepatocellular carcinoma; KPNB1: karyopherin subunit beta 1; PRKN: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; TFEB: transcription factor EB; TMX2: thioredoxin related transmembrane protein 2; VDAC2: voltage dependent anion channel 2; VDAC3: voltage dependent anion channel 3; WB: western blot.
Collapse
Affiliation(s)
- Weiyu Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Yao Tang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Pengfei Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Yutong Chen
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Abdominal Oncology, Cancer Center of The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Zhijie Xu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Chunhui Qi
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Hongbin Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Ruiyang Liu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Haorui Qin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Haoying Ke
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Caini Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Fuyuan Xu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Pengfei Pang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Zhiju Zhao
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Hong Shan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Fei Xiao
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Kashi Guangdong Institute of Science and Technology, The First People’s Hospital of Kashi, Kashi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
100
|
Zhang W, Wang Y, Chen L, Chen H, Qi H, Zheng Y, Du Y, Zhang L, Wang T, Li Q. Dihydroartemisinin suppresses glioma growth by repressing ERRα-mediated mitochondrial biogenesis. Mol Cell Biochem 2024; 479:2809-2825. [PMID: 38072894 DOI: 10.1007/s11010-023-04892-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/31/2023] [Indexed: 10/06/2024]
Abstract
Malignant gliomas are an exceptionally lethal form of cancer with limited treatment options. Dihydroartemisinin (DHA), a sesquiterpene lactone antimalarial compound, has demonstrated therapeutic effects in various solid tumors. In our study, we aimed to investigate the mechanisms underlying the anticancer effects of DHA in gliomas. To explore the therapeutic and molecular mechanisms of DHA, we employed various assays, including cell viability, flow cytometry, mitochondrial membrane potential, glucose uptake and glioma xenograft models. Our data demonstrated that DHA significantly inhibited glioma cell proliferation in both temozolomide-resistant cells and glioma stem-like cells. We found that DHA-induced apoptosis occurred via the mitochondria-mediated pathway by initiating mitochondrial dysfunction before promoting apoptosis. Moreover, we discovered that DHA treatment substantially reduced the expression of the mitochondrial biogenesis-related gene, ERRα, in glioma cells. And the ERRα pathway is a critical target in treating glioma with DHA. Our results also demonstrated that the combination of DHA and temozolomide synergistically inhibited the proliferation of glioma cells. In vivo, DHA treatment remarkably extended survival time in mice bearing orthotopic glioblastoma xenografts. Thus, our findings suggest that DHA has a novel role in modulating cancer cell metabolism and suppressing glioma progression by activating the ERRα-regulated mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Yan Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Haifei Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Yong Zheng
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, People's Republic of China
| | - Yongli Du
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, People's Republic of China
| | - Liudi Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|